Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35042797

ABSTRACT

Srs2 is a superfamily 1 (SF1) helicase that participates in several pathways necessary for the repair of damaged DNA. Srs2 regulates formation of early homologous recombination (HR) intermediates by actively removing the recombinase Rad51 from single-stranded DNA (ssDNA). It is not known whether and how Srs2 itself is down-regulated to allow for timely HR progression. Rad54 and Rdh54 are two closely related superfamily 2 (SF2) motor proteins that promote the formation of Rad51-dependent recombination intermediates. Rad54 and Rdh54 bind tightly to Rad51-ssDNA and act downstream of Srs2, suggesting that they may affect the ability of Srs2 to dismantle Rad51 filaments. Here, we used DNA curtains to determine whether Rad54 and Rdh54 alter the ability of Srs2 to disrupt Rad51 filaments. We show that Rad54 and Rdh54 act synergistically to greatly restrict the antirecombinase activity of Srs2. Our findings suggest that Srs2 may be accorded only a limited time window to act and that Rad54 and Rdh54 fulfill a role of prorecombinogenic licensing factors.


Subject(s)
DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Topoisomerases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage/physiology , DNA Helicases/physiology , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/physiology , DNA Topoisomerases/physiology , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Homologous Recombination/genetics , Protein Binding/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology
2.
Genes (Basel) ; 12(9)2021 09 08.
Article in English | MEDLINE | ID: mdl-34573372

ABSTRACT

Homologous recombination (HR) is a mechanism conserved from bacteria to humans essential for the accurate repair of DNA double-stranded breaks, and maintenance of genome integrity. In eukaryotes, the key DNA transactions in HR are catalyzed by the Rad51 recombinase, assisted by a host of regulatory factors including mediators such as Rad52 and Rad51 paralogs. Rad51 paralogs play a crucial role in regulating proper levels of HR, and mutations in the human counterparts have been associated with diseases such as cancer and Fanconi Anemia. In this review, we focus on the Saccharomyces cerevisiae Rad51 paralog complex Rad55-Rad57, which has served as a model for understanding the conserved role of Rad51 paralogs in higher eukaryotes. Here, we discuss the results from early genetic studies, biochemical assays, and new single-molecule observations that have together contributed to our current understanding of the molecular role of Rad55-Rad57 in HR.


Subject(s)
Adenosine Triphosphatases/physiology , DNA Repair Enzymes/physiology , DNA Repair/physiology , DNA-Binding Proteins/physiology , Homologous Recombination , Saccharomyces cerevisiae Proteins/physiology , Gene Expression Regulation, Fungal , Multiprotein Complexes , Mutation , Saccharomyces cerevisiae/genetics , Single Molecule Imaging
3.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576232

ABSTRACT

Neuroblastoma, the most common extra-cranial solid tumor of early childhood, is one of the major therapeutic challenges in child oncology: it is highly heterogenic at a genetic, biological, and clinical level. The high-risk cases have one of the least favorable outcomes amongst pediatric tumors, and the mortality rate is still high, regardless of the use of intensive multimodality therapies. Here, we observed that neuroblastoma cells display an increased expression of Cockayne Syndrome group B (CSB), a pleiotropic protein involved in multiple functions such as DNA repair, transcription, mitochondrial homeostasis, and cell division, and were recently found to confer cell robustness when they are up-regulated. In this study, we demonstrated that RNAi-mediated suppression of CSB drastically impairs tumorigenicity of neuroblastoma cells by hampering their proliferative, clonogenic, and invasive capabilities. In particular, we observed that CSB ablation induces cytokinesis failure, leading to caspases 9 and 3 activation and, subsequently, to massive apoptotic cell death. Worthy of note, a new frontier in cancer treatment, already proved to be successful, is cytokinesis-failure-induced cell death. In this context, CSB ablation seems to be a new and promising anticancer strategy for neuroblastoma therapy.


Subject(s)
Cytokinesis/physiology , DNA Helicases/physiology , DNA Repair Enzymes/physiology , Neuroblastoma/metabolism , Poly-ADP-Ribose Binding Proteins/physiology , RNA Interference , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Centrosome , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , DNA Repair Enzymes/genetics , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Spindle Apparatus
4.
Nucleic Acids Res ; 49(5): 2418-2434, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33590097

ABSTRACT

Cockayne syndrome (CS) is a segmental premature aging syndrome caused primarily by defects in the CSA or CSB genes. In addition to premature aging, CS patients typically exhibit microcephaly, progressive mental and sensorial retardation and cutaneous photosensitivity. Defects in the CSB gene were initially thought to primarily impair transcription-coupled nucleotide excision repair (TC-NER), predicting a relatively consistent phenotype among CS patients. In contrast, the phenotypes of CS patients are pleiotropic and variable. The latter is consistent with recent work that implicates CSB in multiple cellular systems and pathways, including DNA base excision repair, interstrand cross-link repair, transcription, chromatin remodeling, RNAPII processing, nucleolin regulation, rDNA transcription, redox homeostasis, and mitochondrial function. The discovery of additional functions for CSB could potentially explain the many clinical phenotypes of CSB patients. This review focuses on the diverse roles played by CSB in cellular pathways that enhance genome stability, providing insight into the molecular features of this complex premature aging disease.


Subject(s)
DNA Helicases/physiology , DNA Repair Enzymes/physiology , Poly-ADP-Ribose Binding Proteins/physiology , Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Helicases/chemistry , DNA Repair , DNA Repair Enzymes/chemistry , DNA, Ribosomal/biosynthesis , Gene Expression Regulation , Humans , Mitochondria/genetics , Mitochondria/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , RNA Polymerase II/metabolism , Transcription, Genetic
5.
Nucleic Acids Res ; 48(9): 4960-4975, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32232411

ABSTRACT

G-quadruplexes represent unique roadblocks to DNA replication, which tends to stall at these secondary structures. Although G-quadruplexes can be found throughout the genome, telomeres, due to their G-richness, are particularly predisposed to forming these structures and thus represent difficult-to-replicate regions. Here, we demonstrate that exonuclease 1 (EXO1) plays a key role in the resolution of, and replication through, telomeric G-quadruplexes. When replication forks encounter G-quadruplexes, EXO1 resects the nascent DNA proximal to these structures to facilitate fork progression and faithful replication. In the absence of EXO1, forks accumulate at stabilized G-quadruplexes and ultimately collapse. These collapsed forks are preferentially repaired via error-prone end joining as depletion of EXO1 diverts repair away from error-free homology-dependent repair. Such aberrant repair leads to increased genomic instability, which is exacerbated at chromosome termini in the form of dysfunction and telomere loss.


Subject(s)
DNA Repair Enzymes/physiology , DNA Replication , Exodeoxyribonucleases/physiology , G-Quadruplexes , Telomere/chemistry , Aminoquinolines/pharmacology , Cell Line , DNA End-Joining Repair , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , G-Quadruplexes/drug effects , Gene Knockout Techniques , HeLa Cells , Humans , Neoplasms/metabolism , Neoplasms/mortality , Picolinic Acids/pharmacology , Prognosis
6.
Mutagenesis ; 35(1): 129-149, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31858150

ABSTRACT

Alterations of DNA repair enzymes and consequential triggering of aberrant DNA damage response (DDR) pathways are thought to play a pivotal role in genomic instabilities associated with cancer development, and are further thought to be important predictive biomarkers for therapy using the synthetic lethality paradigm. However, novel unpredicted perspectives are emerging from the identification of several non-canonical roles of DNA repair enzymes, particularly in gene expression regulation, by different molecular mechanisms, such as (i) non-coding RNA regulation of tumour suppressors, (ii) epigenetic and transcriptional regulation of genes involved in genotoxic responses and (iii) paracrine effects of secreted DNA repair enzymes triggering the cell senescence phenotype. The base excision repair (BER) pathway, canonically involved in the repair of non-distorting DNA lesions generated by oxidative stress, ionising radiation, alkylation damage and spontaneous or enzymatic deamination of nucleotide bases, represents a paradigm for the multifaceted roles of complex DDR in human cells. This review will focus on what is known about the canonical and non-canonical functions of BER enzymes related to cancer development, highlighting novel opportunities to understand the biology of cancer and representing future perspectives for designing new anticancer strategies. We will specifically focus on APE1 as an example of a pleiotropic and multifunctional BER protein.


Subject(s)
DNA Repair Enzymes/physiology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Neoplasms/enzymology , DNA/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
7.
Cytogenet Genome Res ; 158(1): 25-31, 2019.
Article in English | MEDLINE | ID: mdl-31055587

ABSTRACT

Diagnosing a complex genetic syndrome and correctly assigning the concomitant phenotypic traits to a well-defined clinical form is often a medical challenge. In this work, we report the analysis of a family with complex phenotypes, including microcephaly, intellectual disability, dysmorphic features, and polydactyly in the proband, with the aim of adding new aspects for obtaining a clear diagnosis. We performed array-comparative genomic hybridization and quantitative reverse transcriptase PCR (qRT-PCR) analyses. We identified a deletion of chromosome 20p12.1 involving the macrodomain containing 2/mono-ADP ribosylhydrolase 2 gene (MACROD2) in several members of the family. This gene is actually not associated with a specific syndrome but with congenital anomalies of multiple organs. qRT-PCR showed higher levels of a MACROD2 mRNA isoform in the individuals carrying the deletion. Our results, together with other data reported in the literature, support the hypothesis that the deletion in MACROD2 can affect correct embryonic development and that the presence of another associated event, such as epigenetic modifications at the MACROD2 locus, can influence the level of severity of the pathology.


Subject(s)
Abnormalities, Multiple/genetics , DNA Repair Enzymes/genetics , Hydrolases/genetics , Intellectual Disability/genetics , Kidney/abnormalities , Microcephaly/genetics , Pancreas/abnormalities , Polydactyly/genetics , Sequence Deletion , Adult , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, Pair 20/ultrastructure , Comparative Genomic Hybridization , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/physiology , Embryonic Development/genetics , Female , Humans , Hydrolases/deficiency , Hydrolases/physiology , Male , Pedigree , Phenotype , Psychomotor Disorders/genetics
8.
Nat Struct Mol Biol ; 25(10): 971-980, 2018 10.
Article in English | MEDLINE | ID: mdl-30291363

ABSTRACT

The Ku70-Ku80 (Ku) heterodimer binds rapidly and tightly to the ends of DNA double-strand breaks and recruits factors of the non-homologous end-joining (NHEJ) repair pathway through molecular interactions that remain unclear. We have determined crystal structures of the Ku-binding motifs (KBM) of the NHEJ proteins APLF (A-KBM) and XLF (X-KBM) bound to a Ku-DNA complex. The two KBM motifs bind remote sites of the Ku80 α/ß domain. The X-KBM occupies an internal pocket formed by an unprecedented large outward rotation of the Ku80 α/ß domain. We observe independent recruitment of the APLF-interacting protein XRCC4 and of XLF to laser-irradiated sites via binding of A- and X-KBMs, respectively, to Ku80. Finally, we show that mutation of the X-KBM and A-KBM binding sites in Ku80 compromises both the efficiency and accuracy of end joining and cellular radiosensitivity. A- and X-KBMs may represent two initial anchor points to build the intricate interaction network required for NHEJ.


Subject(s)
DNA End-Joining Repair , DNA Repair Enzymes/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-Binding Proteins/chemistry , Ku Autoantigen/chemistry , Poly-ADP-Ribose Binding Proteins/chemistry , Conserved Sequence , Crystallography, X-Ray , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Humans , Ku Autoantigen/metabolism , Ku Autoantigen/physiology , Models, Molecular , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/physiology , Protein Domains
9.
PLoS Genet ; 14(10): e1007719, 2018 10.
Article in English | MEDLINE | ID: mdl-30312299

ABSTRACT

The Fragile X-related disorders (FXDs) are Repeat Expansion Diseases resulting from an expansion of a CGG-repeat tract at the 5' end of the FMR1 gene. The mechanism responsible for this unusual mutation is not fully understood. We have previously shown that mismatch repair (MMR) complexes, MSH2/MSH3 (MutSß) and MSH2/MSH6 (MutSα), together with Polß, a DNA polymerase important for base excision repair (BER), are important for expansions in a mouse model of these disorders. Here we show that MLH1/MLH3 (MutLγ), a protein complex that can act downstream of MutSß in MMR, is also required for all germ line and somatic expansions. However, exonuclease I (EXO1), which acts downstream of MutL proteins in MMR, is not required. In fact, a null mutation in Exo1 results in more extensive germ line and somatic expansions than is seen in Exo1+/+ animals. Furthermore, mice homozygous for a point mutation (D173A) in Exo1 that eliminates its nuclease activity but retains its native conformation, shows a level of expansion that is intermediate between Exo1+/+ and Exo1-/- animals. Thus, our data suggests that expansion of the FX repeat in this mouse model occurs via a MutLγ-dependent, EXO1-independent pathway, with EXO1 protecting against expansion both in a nuclease-dependent and a nuclease-independent manner. Our data thus have implications for the expansion mechanism and add to our understanding of the genetic factors that may be modifiers of expansion risk in humans.


Subject(s)
DNA Repair Enzymes/genetics , Exodeoxyribonucleases/genetics , Fragile X Syndrome/genetics , MutL Proteins/genetics , Animals , DNA Mismatch Repair/genetics , DNA Mismatch Repair/physiology , DNA Repair , DNA Repair Enzymes/physiology , Disease Models, Animal , Exodeoxyribonucleases/physiology , Fragile X Mental Retardation Protein/genetics , Genomic Instability , Mice , Mice, Inbred C57BL , Mice, Knockout , MutL Protein Homolog 1/metabolism , MutL Proteins/metabolism , Mutation , Trinucleotide Repeat Expansion/genetics
10.
Nucleic Acids Res ; 46(20): 10888-10904, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30304478

ABSTRACT

Nucleotides in the free pool are more susceptible to nonenzymatic methylation than those protected in the DNA double helix. Methylated nucleotides like O6-methyl-dGTP can be mutagenic and toxic if incorporated into DNA. Removal of methylated nucleotides from the nucleotide pool may therefore be important to maintain genome integrity. We show that MutT homologue 1 (MTH1) efficiently catalyzes the hydrolysis of O6-methyl-dGTP with a catalytic efficiency similar to that for 8-oxo-dGTP. O6-methyl-dGTP activity is exclusive to MTH1 among human NUDIX proteins and conserved through evolution but not found in bacterial MutT. We present a high resolution crystal structure of human and zebrafish MTH1 in complex with O6-methyl-dGMP. By microinjecting fertilized zebrafish eggs with O6-methyl-dGTP and inhibiting MTH1 we demonstrate that survival is dependent on active MTH1 in vivo. O6-methyl-dG levels are higher in DNA extracted from zebrafish embryos microinjected with O6-methyl-dGTP and inhibition of O6-methylguanine-DNA methyl transferase (MGMT) increases the toxicity of O6-methyl-dGTP demonstrating that O6-methyl-dGTP is incorporated into DNA. MTH1 deficiency sensitizes human cells to the alkylating agent Temozolomide, a sensitization that is more pronounced upon MGMT inhibition. These results expand the cellular MTH1 function and suggests MTH1 also is important for removal of methylated nucleotides from the nucleotide pool.


Subject(s)
DNA Repair Enzymes/physiology , Deoxyguanine Nucleotides/chemistry , Phosphoric Monoester Hydrolases/physiology , Animals , Catalytic Domain , Crystallography, X-Ray , DNA Modification Methylases/chemistry , DNA Repair Enzymes/chemistry , Dogs , Escherichia coli/genetics , HL-60 Cells , Humans , Hydrolysis , Kinetics , Mice , Nucleotides , Phosphoric Monoester Hydrolases/chemistry , Pyrophosphatases/chemistry , Species Specificity , Swine , Temozolomide/pharmacology , Tumor Suppressor Proteins/chemistry , Zebrafish
11.
Curr Opin Struct Biol ; 53: 22-31, 2018 12.
Article in English | MEDLINE | ID: mdl-29656238

ABSTRACT

Terminal deoxynucleotidyltransferase (TdT) is a member of the polX family which is involved in DNA repair. It has been known for years as an untemplated DNA polymerase used during V(D)J recombination to generate diversity at the CDR3 region of immunoglobulins and T-cell receptors. Recently, however, TdT was crystallized in the presence of a complete DNA synapsis made of two double-stranded DNA (dsDNA), each with a 3' protruding end, and overlapping with only one micro-homology base-pair, thus giving structural insight for the first time into DNA synthesis across strands. It was subsequently shown that TdT indeed has an in trans template-dependent activity in the presence of an excess of the downstream DNA duplex. A possible biological role of this dual activity is discussed.


Subject(s)
DNA Nucleotidylexotransferase , Animals , DNA Nucleotidylexotransferase/chemistry , DNA Nucleotidylexotransferase/metabolism , DNA Nucleotidylexotransferase/physiology , DNA Repair , DNA Repair Enzymes/physiology , DNA-Directed DNA Polymerase/chemistry , Humans , V(D)J Recombination
12.
Bull Cancer ; 104(11): 981-987, 2017 Nov.
Article in French | MEDLINE | ID: mdl-29132682

ABSTRACT

The identification of DNA repair biomarkers is of paramount importance. Indeed, it is the first step in the process of modulating radiosensitivity and radioresistance. Unlike tools of detection and measurement of DNA damage, DNA repair biomarkers highlight the variations of DNA damage responses, depending on the dose and the dose rate. The aim of the present review is to describe the main biomarkers of radiation-induced DNA repair. We will focus on double strand breaks (DSB), because of their major role in radiation-induced cell death. The most important DNA repair biomarkers are DNA damage signaling proteins, with ATM, DNA-PKcs, 53BP1 and γ-H2AX. They can be analyzed either using immunostaining, or using lived cell imaging. However, to date, these techniques are still time and money consuming. The development of "omics" technologies should lead the way to new (and usable in daily routine) DNA repair biomarkers.


Subject(s)
Biomarkers/analysis , DNA Repair , DNA/radiation effects , Ataxia Telangiectasia Mutated Proteins/physiology , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair Enzymes/adverse effects , DNA Repair Enzymes/physiology , DNA-Activated Protein Kinase/physiology , Dose-Response Relationship, Radiation , Histones/physiology , Humans , Nuclear Proteins/physiology , Radiation Tolerance , Radiotherapy , Recombinational DNA Repair , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/physiology
13.
Cancer Res ; 77(19): 5327-5338, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28819025

ABSTRACT

Hypomorphic mutations in the genes encoding the MRE11/RAD50/NBS1 (MRN) DNA repair complex lead to cancer-prone syndromes. MRN binds DNA double-strand breaks, where it functions in repair and triggers cell-cycle checkpoints via activation of the ataxia-telangiectasia mutated kinase. To gain understanding of MRN in cancer, we engineered mice with B lymphocytes lacking MRN, or harboring MRN in which MRE11 lacks nuclease activities. Both forms of MRN deficiency led to hallmarks of cancer, including oncogenic translocations involving c-Myc and the immunoglobulin locus. These preneoplastic B lymphocytes did not progress to detectable B lineage lymphoma, even in the absence of p53. Moreover, Mre11 deficiencies prevented tumorigenesis in a mouse model strongly predisposed to spontaneous B-cell lymphomas. Our findings indicate that MRN cannot be considered a standard tumor suppressor and instead imply that nuclease activities of MRE11 are required for oncogenesis. Inhibition of MRE11 nuclease activity increased DNA damage and selectively induced apoptosis in cells overexpressing oncogenes, suggesting MRE11 serves an important role in countering oncogene-induced replication stress. Thus, MRE11 may offer a target for cancer therapeutic development. More broadly, our work supports the idea that subtle enhancements of endogenous genome instability can exceed the tolerance of cancer cells and be exploited for therapeutic ends. Cancer Res; 77(19); 5327-38. ©2017 AACR.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , DNA Repair Enzymes/physiology , DNA Replication , DNA-Binding Proteins/physiology , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins c-myc/metabolism , ATP-Binding Cassette Transporters/physiology , Acid Anhydride Hydrolases , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/metabolism , Cell Cycle Proteins/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Repair , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Genomic Instability , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , MRE11 Homologue Protein , Mice , Mutation , Nuclear Proteins/physiology , Oncogenes , Proto-Oncogene Proteins c-myc/genetics
14.
Proc Natl Acad Sci U S A ; 114(23): 6010-6015, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533382

ABSTRACT

Human exonuclease 1 (hExo1) is a member of the RAD2/XPG structure-specific 5'-nuclease superfamily. Its dominant, processive 5'-3' exonuclease and secondary 5'-flap endonuclease activities participate in various DNA repair, recombination, and replication processes. A single active site processes both recessed ends and 5'-flap substrates. By initiating enzyme reactions in crystals, we have trapped hExo1 reaction intermediates that reveal structures of these substrates before and after their exo- and endonucleolytic cleavage, as well as structures of uncleaved, unthreaded, and partially threaded 5' flaps. Their distinctive 5' ends are accommodated by a small, mobile arch in the active site that binds recessed ends at its base and threads 5' flaps through a narrow aperture within its interior. A sequence of successive, interlocking conformational changes guides the two substrate types into a shared reaction mechanism that catalyzes their cleavage by an elaborated variant of the two-metal, in-line hydrolysis mechanism. Coupling of substrate-dependent arch motions to transition-state stabilization suppresses inappropriate or premature cleavage, enhancing processing fidelity. The striking reduction in flap conformational entropy is catalyzed, in part, by arch motions and transient binding interactions between the flap and unprocessed DNA strand. At the end of the observed reaction sequence, hExo1 resets without relinquishing DNA binding, suggesting a structural basis for its processivity.


Subject(s)
DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Biocatalysis , Catalytic Domain/physiology , Crystallography, X-Ray , DNA/chemistry , DNA Repair , DNA Repair Enzymes/physiology , DNA-Binding Proteins/chemistry , Endonucleases/metabolism , Exodeoxyribonucleases/physiology , Humans , Hydrolysis , Protein Conformation , Substrate Specificity/physiology
15.
Nucleic Acids Res ; 45(10): 6238-6251, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28453785

ABSTRACT

Non-homologous end joining (NHEJ) repairs DNA double strand breaks in non-cycling eukaryotic cells. NHEJ relies on polynucleotide kinase/phosphatase (PNKP), which generates 5΄-phosphate/3΄-hydroxyl DNA termini that are critical for ligation by the NHEJ DNA ligase, LigIV. PNKP and LigIV require the NHEJ scaffolding protein, XRCC4. The PNKP FHA domain binds to the CK2-phosphorylated XRCC4 C-terminal tail, while LigIV uses its tandem BRCT repeats to bind the XRCC4 coiled-coil. Yet, the assembled PNKP-XRCC4-LigIV complex remains uncharacterized. Here, we report purification and characterization of a recombinant PNKP-XRCC4-LigIV complex. We show that the stable binding of PNKP in this complex requires XRCC4 phosphorylation and that only one PNKP protomer binds per XRCC4 dimer. Small angle X-ray scattering (SAXS) reveals a flexible multi-state complex that suggests that both the PNKP FHA and catalytic domains contact the XRCC4 coiled-coil and LigIV BRCT repeats. Hydrogen-deuterium exchange indicates protection of a surface on the PNKP phosphatase domain that may contact XRCC4-LigIV. A mutation on this surface (E326K) causes the hereditary neuro-developmental disorder, MCSZ. This mutation impairs PNKP recruitment to damaged DNA in human cells and provides a possible disease mechanism. Together, this work unveils multipoint contacts between PNKP and XRCC4-LigIV that regulate PNKP recruitment and activity within NHEJ.


Subject(s)
DNA End-Joining Repair/physiology , DNA Ligase ATP/physiology , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Catalytic Domain , DNA Damage , DNA Ligase ATP/chemistry , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA-Binding Proteins/chemistry , Deuterium/metabolism , Developmental Disabilities/genetics , Humans , Mass Spectrometry , Microcephaly/genetics , Models, Molecular , Multiprotein Complexes , Mutation, Missense , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Point Mutation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle , Seizures/genetics , Syndrome , X-Ray Diffraction
16.
Yakugaku Zasshi ; 137(3): 293-300, 2017.
Article in Japanese | MEDLINE | ID: mdl-28250323

ABSTRACT

8-Oxo-2'-deoxyguanosine (8-oxo-dG) is a representative of nucleoside damage, which is generated by the reaction of the 8 position of dG with reactive oxygen species. Abundant 8-oxo-dG in DNA exhibits genotoxicity and has been linked to aging and disease, such as cancer. As the metabolism of cancer cells is much faster than that of normal cells, the oxidized product of the oligonucleotides and the nucleotide pool produces 8-oxo-dG and 8-oxo-2'-deoxyguanosine triphosphate (8-oxo-dGTP), respectively. Human oxoguanine glycosylase (hOGG1) shows base excision activity for 8-oxo-dG in duplex DNA. On the other hand, human mutT homologue protein (hMTH1, also known as NUDT1) is important for oxidized nucleotide removal including 8-oxo-dGTP, and it is reported that the presence of hMTH1 is not essential for normal cells but is required for the survival of cancer cells. Therefore, we designed and synthesized 8-halogenated 7-deaza-2'-deoxyguanosine triphosphate (8-halo-7-deaza-dGTP) derivatives as mimics of 8-oxo-dGTP in order to interact with hMTH1. The 8-halo-7-deaza-dGTP derivatives were poor substrates for but strong binders to hMTH1. Interestingly, they exhibited strong competitive inhibition of hMTH1 in the hydrolysis of 8-oxo-dGTP. This inhibitory effect is caused by the slower rate of hydrolysis due to possible small enzyme structural changes. Although the detailed inhibition mechanism of the hydrolysis activity of hMTH1 is unknown, this result is the first to demonstrate the potential of nucleoside triphosphate derivatives as antitumor agents.


Subject(s)
DNA/genetics , Deoxyadenosines , Deoxyguanine Nucleotides , Nucleosides , Tubercidin/analogs & derivatives , Antineoplastic Agents , DNA Glycosylases/physiology , DNA Repair Enzymes/physiology , Humans , Hydrolysis , Neoplasms/genetics , Neoplasms/metabolism , Oxidation-Reduction , Phosphoric Monoester Hydrolases/physiology , Reactive Oxygen Species , Tubercidin/chemical synthesis
17.
Bioessays ; 39(3)2017 03.
Article in English | MEDLINE | ID: mdl-28133776

ABSTRACT

Non-homologous end-joining (NHEJ) is the dominant means of repairing chromosomal DNA double strand breaks (DSBs), and is essential in human cells. Fifteen or more proteins can be involved in the detection, signalling, synapsis, end-processing and ligation events required to repair a DSB, and must be assembled in the confined space around the DNA ends. We review here a number of interaction points between the core NHEJ components (Ku70, Ku80, DNA-PKcs, XRCC4 and Ligase IV) and accessory factors such as kinases, phosphatases, polymerases and structural proteins. Conserved protein-protein interaction sites such as Ku-binding motifs (KBMs), XLF-like motifs (XLMs), FHA and BRCT domains illustrate that different proteins compete for the same binding sites on the core machinery, and must be spatially and temporally regulated. We discuss how post-translational modifications such as phosphorylation, ADP-ribosylation and ubiquitinylation may regulate sequential steps in the NHEJ pathway or control repair at different types of DNA breaks.


Subject(s)
DNA End-Joining Repair , Amino Acid Sequence , Animals , DNA Breaks, Double-Stranded , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/physiology , Humans , Protein Interaction Domains and Motifs , Protein Interaction Maps
18.
PLoS One ; 11(11): e0165509, 2016.
Article in English | MEDLINE | ID: mdl-27824946

ABSTRACT

The silencing of the tumor suppressor gene O-6-methylguanine-DNA methyltransferase (MGMT) by promoter methylation commonly occurs in human cancers. The relationship between MGMT promoter methylation and gastric cancer (GC) remains inconsistent. This study aimed to evaluate the potential value of MGMT promoter methylation in GC patients. Electronic databases were searched to identify eligible studies. The pooled odds ratio (OR) and the corresponding 95% confidence interval (95% CI) were used to evaluate the effects of MGMT methylation on GC risk and clinicopathological characteristics. In total, 31 eligible studies including 2988 GC patients and 2189 nonmalignant controls were involved in meta-analysis. In the pooled analysis, MGMT promoter methylation was significantly associated with GC risk (OR = 3.34, P < 0.001) and substantial heterogeneity (P < 0.001). Meta-regression and subgroup analyses based on the testing method, sample material and ethnicity failed to explain the sources of heterogeneity. Interestingly, MGMT methylation showed a trend associated with gender, and methylation is lower in males compared with females (OR = 0.76, 95% CI = 0.56-1.03). We did not find a significant association in relation to tumor types, clinical stage, age status or H. pylori status in cancer (all P > 0.1). MGMT promoter methylation may be correlated with the prognosis of GCs in disease free survival (DFS) or overall survival (OS) for univariate analysis. MGMT promoter methylation may play a crucial role in the carcinogenesis and prognosis of GC. MGMT methylation was not correlated with tumor types, clinical stage, age status, H. pylori status. However, the result of the association of MGMT methylation and gender should be considered with caution.


Subject(s)
DNA Methylation , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Promoter Regions, Genetic/physiology , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Female , Humans , Male , Sex Factors , Stomach Neoplasms/etiology
20.
Oncotarget ; 7(34): 54430-54444, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27303920

ABSTRACT

Ionizing radiation (IR) induces highly cytotoxic double-strand breaks (DSBs) and also clustered oxidized bases in mammalian genomes. Base excision repair (BER) of bi-stranded oxidized bases could generate additional DSBs as repair intermediates in the vicinity of direct DSBs, leading to loss of DNA fragments. This could be avoided if DSB repair via DNA-PK-mediated nonhomologous end joining (NHEJ) precedes BER initiated by NEIL1 and other DNA glycosylases (DGs). Here we show that DNA-PK subunit Ku inhibits DGs via direct interaction. The scaffold attachment factor (SAF)-A, (also called hnRNP-U), phosphorylated at Ser59 by DNA-PK early after IR treatment, is linked to transient release of chromatin-bound NEIL1, thus preventing BER. SAF-A is subsequently dephosphorylated. Ku inhibition of DGs in vitro is relieved by unphosphorylated SAF-A, but not by the phosphomimetic Asp59 mutant. We thus propose that SAF-A, in concert with Ku, temporally regulates base damage repair in irradiated cell genome.


Subject(s)
DNA Repair , Heterogeneous-Nuclear Ribonucleoprotein U/physiology , Ku Autoantigen/physiology , Radiation Injuries/etiology , DNA Breaks, Double-Stranded , DNA Glycosylases/physiology , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , HEK293 Cells , Humans , Phosphorylation , Radiation Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL
...